US20210009592A1 - Organic compounds - Google Patents

Organic compounds Download PDF

Info

Publication number
US20210009592A1
US20210009592A1 US17/040,892 US201917040892A US2021009592A1 US 20210009592 A1 US20210009592 A1 US 20210009592A1 US 201917040892 A US201917040892 A US 201917040892A US 2021009592 A1 US2021009592 A1 US 2021009592A1
Authority
US
United States
Prior art keywords
disorder
compound
disorders
compounds
compound according
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/040,892
Other languages
English (en)
Inventor
Peng Li
Qiang Zhang
Robert Davis
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Intra Cellular Therapies Inc
Original Assignee
Intra Cellular Therapies Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Intra Cellular Therapies Inc filed Critical Intra Cellular Therapies Inc
Priority to US17/040,892 priority Critical patent/US20210009592A1/en
Priority claimed from PCT/US2019/023350 external-priority patent/WO2019183341A1/en
Publication of US20210009592A1 publication Critical patent/US20210009592A1/en
Assigned to INTRA-CELLULAR THERAPIES, INC. reassignment INTRA-CELLULAR THERAPIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DAVIS, ROBERT, LI, PENG, ZHANG, QIANG
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/16Peri-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • the invention relates to particular deuterated substituted heterocycle fused gamma-carbolines, in free, solid, pharmaceutically acceptable salt and/or substantially pure form as described herein, pharmaceutical compositions thereof, and methods of use in the treatment of diseases involving the 5-HT 2A receptor, the serotonin transporter (SERT), pathways involving dopamine D 1 and/or D 2 receptor signaling systems, and/or the ⁇ -opioid receptor, e.g., diseases or disorders such as anxiety, psychosis, schizophrenia, sleep disorders, sexual disorders, migraine, conditions associated with pain (including cephalic pain, neuropathic pain, and as an acute analgesic), fibromyalgia, chronic fatigue, social phobias, gastrointestinal disorders such as dysfunction of the gastrointestinal tract motility and obesity; depression and mood disorders, such as those associated with psychosis or Parkinson's disease; psychosis such as schizophrenia associated with depression; bipolar disorder; drug dependencies, such as opiate dependency and alcohol dependency, drug withdrawal symptoms; obsessive-compuls
  • Substituted heterocycle fused gamma-carbolines are known to be agonists or antagonists of 5-HT 2 receptors, particularly 5-HT 2A receptor, in treating central nervous system disorders.
  • 5-HT 2 receptors particularly 5-HT 2A receptor
  • These compounds have been disclosed in U.S. Pat. Nos. 6,548,493; 7,238,690; 6,552,017; 6,713,471; 7,183,282; U.S. RE39680, and U.S. RE39679, as novel compounds useful for the treatment of disorders associated with 5-HT 2A receptor modulation such as obesity, anxiety, depression, psychosis, schizophrenia, sleep disorders, sexual disorders migraine, conditions associated with cephalic pain, social phobias, gastrointestinal disorders such as dysfunction of the gastrointestinal tract motility, and obesity.
  • Patent Publications 2010/113781 and 2004/209864 also disclose methods of making substituted heterocycle fused gamma-carbolines and uses of these gamma-carbolines as serotonin agonists and antagonists useful for the control and prevention of central nervous system disorders such as addictive behavior and sleep disorders.
  • US 2011/071080 discloses use of particular substituted heterocycle fused gamma-carbolines for the treatment of a combination of psychosis and depressive disorders as well as sleep, depressive and/or mood disorders in patients with psychosis or Parkinson's disease.
  • this patent application discloses and claims use of these compounds at a low dose to selectively antagonize 5-HT 2A receptors without affecting or minimally affecting dopamine D 2 receptors, thereby useful for the treatment of sleep disorders without the side effects associated with high occupancy of the dopamine D 2 pathways or side effects of other pathways (e.g., GABA A receptors) associated with conventional sedative-hypnotic agents (e.g., benzodiazepines) including but not limited to the development of drug dependency, muscle hypotonia, weakness, headache, blurred vision, vertigo, nausea, vomiting, epigastric distress, diarrhea, joint pains, and chest pains.
  • US 2011/112105 also discloses of methods of preparing toluenesulfonic acid addition salt crystals of these substituted heterocycle fused gamma-carbolines.
  • OCD Obsessive-compulsive disorder
  • OCD Obsessive-compulsive disorder
  • SSRIs selective serotonin reuptake inhibitors
  • a second line of therapy is with antipsychotic agents, such as clomipramine, risperidone, quetiapine and olanzapine.
  • Compounds of Formula A and B are a potent serotonin 5-HT 2A receptor antagonists and mu-opiate receptor partial agonists. These compounds also interact with dopamine receptors, particular the dopamine D1 receptors.
  • the Compounds of Formula A and B and their analogs are useful for the treatment or prophylaxis of central nervous system disorders, but there is a need in the art for analogs, such as isotopic analogs, of the Compounds of Formula A and B that when administered to a patient can provide for improved therapeutic concentrations or improved pharmacokinetic distribution or dynamics of these compounds.
  • the present disclosure fills this need by providing Compounds of Formula I and II, et seq., which are deuterated analogs of the Compounds of Formula A and B.
  • the Compounds of the present disclosure may be particularly suited for formulation as long-acting or extended-release compositions that when administered to a patient can provide for improved therapeutic amounts concentrations of the compounds A and B and their analogs over an extended period of time.
  • the present disclosure relates to a compound (Compound I) of Formula I:
  • the present disclosure provides each of the foregoing Compound I or 1.1-1.25, (hereinafter collectively “Compounds of Formulas I et seq.” or “compounds of the disclosure”) in pharmaceutically acceptable salt form.
  • the present disclosure provides additional exemplary embodiments of the Compounds of Formulas I et seq., including:
  • the present disclosure provides a pharmaceutical composition (Pharmaceutical Composition 3) comprising a compound according to any one of Compound I or 1.1-1.25, e.g., in admixture with a pharmaceutically acceptable diluent or carrier.
  • a pharmaceutical composition comprising a compound according to any one of Compound I or 1.1-1.25, e.g., in admixture with a pharmaceutically acceptable diluent or carrier.
  • the present disclosure provides additional exemplary embodiments of Pharmaceutical Composition 1, including:
  • the Pharmaceutical Compositions of the present disclosure are for a sustained or delayed release, e.g., depot, formulation.
  • the depot formulation (Depot Formulation 3.3) is the Pharmaceutical Composition of any of 3.1-3.3, preferably in free or pharmaceutically acceptable salt form, and preferably in admixture with a pharmaceutically acceptable diluent or carrier, e.g., providing sustained or delayed release as an injectable depot.
  • the Depot Formulation 3.3 comprises a compound according to any one of Compound I or 1.1-1.25, in free base or pharmaceutically acceptable salt form, optionally in crystal form, wherein the compound has been milled to, or the compound crystallized to, microparticle or nanoparticle size, e.g., particles or crystals having a volume-based particle size (e.g., diameter or Dv50) of 0.5 to 100 microns, for example, for example, 5-30 microns, 10-20 microns, 20-100 microns, 20-50 microns or 30-50 microns.
  • Such particles or crystals may be combined with a suitable pharmaceutically acceptable diluent or carrier, for example water, to form a depot formulation for injection.
  • the depot formulation may be formulated for intramuscular or subcutaneous injection with a dosage of drug suitable for 4 to 6 weeks of treatment.
  • the particles or crystals have a surface area of 0.1 to 5 m 2 /g, for example, 0.5 to 3.3 m 2 /g or from 0.8 to 1.2 m 2 /g.
  • the present disclosure provides Pharmaceutical Composition 3.4, which is Pharmaceutical Composition 3 or any of 3.1-3.3, wherein the Compound of Formulas I et seq. is in a polymeric matrix.
  • the Compound of the present disclosure is dispersed or dissolved within the polymeric matrix.
  • the polymeric matrix comprises standard polymers used in depot formulations such as polymers selected from a polyester of a hydroxyfatty acid and derivatives thereof, or a polymer of an alkyl alpha-cyanoacrylate, a polyalkylene oxalate, a polyortho ester, a polycarbonate, a polyortho-carbonate, a polyamino acid, a hyaluronic acid ester, and mixtures thereof.
  • the polymer is selected from a group consisting of polylactide, poly d,l-lactide, poly glycolide, PLGA 50:50, PLGA 85:15 and PLGA 90:10 polymer.
  • the polymer is selected form poly(glycolic acid), poly-D,L-lactic acid, poly-L-lactic acid, copolymers of the foregoing, poly(aliphatic carboxylic acids), copolyoxalates, polycaprolactone, polydioxanone, poly(ortho carbonates), poly(acetals), poly(lactic acid-caprolactone), polyorthoesters, poly(glycolic acid-caprolactone), polyanhydrides, and natural polymers including albumin, casein, and waxes, such as, glycerol mono- and distearate, and the like.
  • the polymeric matrix comprises poly(d,l-lactide-co-glycolide).
  • compositions 3 and 3.1-3.4 are particularly useful for sustained or delayed release, wherein the Compound of the present disclosure is released upon degradation of the polymeric matrix.
  • These Compositions may be formulated for controlled- and/or sustained-release of the Compounds of the present disclosure (e.g., as a depot composition) over a period of up to 180 days, e.g., from about 14 to about 30 to about 180 days.
  • the polymeric matrix may degrade and release the Compounds of the present disclosure over a period of about 30, about 60 or about 90 days.
  • the polymeric matrix may degrade and release the Compounds of the present disclosure over a period of about 120, or about 180 days.
  • compositions of the present disclosure for example the depot composition of the present disclosure, e.g., Pharmaceutical Composition 3.3 or 3.4, is formulated for administration by injection.
  • the present disclosure provides the Compounds of Formulas I et seq. as hereinbefore described, in an osmotic controlled release oral delivery system (OROS), which is described in WO 2000/35419 (US 2001/0036472) and EP 1 539 115 (U.S. Pub. No. 2009/0202631), the contents of each of which applications are incorporated by reference in their entirety. Therefore in one embodiment of the seventh aspect, the present disclosure provides a pharmaceutical composition or device comprising (a) a gelatin capsule containing a Compound of any of Formulae I et seq.
  • OROS osmotic controlled release oral delivery system
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a gelatin capsule containing a liquid, the Compound of Formulas I et seq. in free or pharmaceutically acceptable salt form or a Pharmaceutical Composition of the Invention, e.g., any of Pharmaceutical Composition 3 or 3.1-3.4, the gelatin capsule being surrounded by a composite wall comprising a barrier layer contacting the external surface of the gelatin capsule, an expandable layer contacting the barrier layer, a semi-permeable layer encompassing the expandable layer, and an exit orifice formed or formable in the wall.
  • a pharmaceutical composition comprising a gelatin capsule containing a liquid, the Compound of Formulas I et seq. in free or pharmaceutically acceptable salt form or a Pharmaceutical Composition of the Invention, e.g., any of Pharmaceutical Composition 3 or 3.1-3.4, the gelatin capsule being surrounded by a composite wall comprising a barrier layer contacting the external surface of the gelatin capsule, an expandable layer contacting the barrier layer, a
  • the invention provides a composition comprising a gelatin capsule containing a liquid, the Compound of Formulas I et seq. in free or pharmaceutically acceptable salt form or a Pharmaceutical Composition of the Invention, e.g., any of Pharmaceutical Composition 3 or 3.1-3.4, the gelatin capsule being surrounded by a composite wall comprising a barrier layer contacting the external surface of the gelatin capsule, an expandable layer contacting the barrier layer, a semipermeable layer encompassing the expandable layer, and an exit orifice formed or formable in the wall, wherein the barrier layer forms a seal between the expandable layer and the environment at the exit orifice.
  • a Pharmaceutical Composition of the Invention e.g., any of Pharmaceutical Composition 3 or 3.1-3.4
  • the invention provides a composition comprising a gelatin capsule containing a liquid, the Compound of Formulas I et seq. in free or pharmaceutically acceptable salt form or a Pharmaceutical Composition of the Invention, e.g., any of Pharmaceutical Composition 3 or 3.1-3.4, the gelatin capsule being surrounded by a barrier layer contacting the external surface of the gelatin capsule, an expandable layer contacting a portion of the barrier layer, a semi-permeable layer encompassing at least the expandable layer, and an exit orifice formed or formable in the dosage form extending from the external surface of the gelatin capsule to the environment of use.
  • the expandable layer may be formed in one or more discrete sections, such as for example, two sections located on opposing sides or ends of the gelatin capsule.
  • the Compound of the present disclosure in the Osmotic-controlled Release Oral Delivery System (i.e., in Pharmaceutical Composition P.1-P.4) is in a liquid formulation, which formulation may be neat, liquid active agent, liquid active agent in a solution, suspension, emulsion or self-emulsifying composition or the like.
  • Osmotic-controlled Release Oral Delivery System composition including characteristics of the gelatin capsule, barrier layer, an expandable layer, a semi-permeable layer; and orifice may be found in WO 2000/35419 (and equivalent US 2001/0036472), the contents of which are incorporated by reference in their entirety.
  • the invention provides a composition or device comprising (a) two or more layers, said two or more layers comprising a first layer and a second layer, said first layer comprises the Compound of Formula I et seq., in free or pharmaceutically acceptable salt form, or a Pharmaceutical Composition as herein before described said second layer comprises a polymer; (b) an outer wall surrounding said two or more layers; and (c) an orifice in said outer wall.
  • a composition or device comprising (a) two or more layers, said two or more layers comprising a first layer and a second layer, said first layer comprises the Compound of Formula I et seq., in free or pharmaceutically acceptable salt form, or a Pharmaceutical Composition as herein before described said second layer comprises a polymer; (b) an outer wall surrounding said two or more layers; and (c) an orifice in said outer wall.
  • composition P.5 preferably utilizes a semi-permeable membrane surrounding a three-layer-core: in these embodiments, the first layer is referred to as a first drug layer and contains low amounts of drug (e.g., the Compound of Formulas I et seq.) and an osmotic agent such as salt, the middle layer referred to as the second drug layer contains higher amounts of drug, excipients and no salt; and the third layer referred to as the push layer contains osmotic agents and no drug. At least one orifice is drilled through the membrane on the first drug layer end of the capsule-shaped tablet. (Pharmaceutical Composition P.6)
  • composition P.5 or P.6 may comprise a membrane defining a compartment, the membrane surrounding an inner protective subcoat, at least one exit orifice formed or formable therein and at least a portion of the membrane being semi-permeable; an expandable layer located within the compartment remote from the exit orifice and in fluid communication with the semi-permeable portion of the membrane; a first drug layer located adjacent the exit orifice; and a second drug layer located within the compartment between the first drug layer and the expandable layer, the drug layers comprising the Compound of the Invention in free or pharmaceutically acceptable salt thereof.
  • different release profiles are obtained. It is imperative to identify the optimum viscosity for each layer. In the present invention, viscosity is modulated by addition of salt, sodium chloride.
  • the delivery profile from the core is dependent on the weight, formulation and thickness of each of the drug layers.
  • the invention provides Pharmaceutical Composition P.7 wherein the first drug layer comprising salt and the second drug layer containing no salt.
  • Pharmaceutical Composition P.5-P.7 may optionally comprise a flow-promoting layer between the membrane and the drug layers.
  • compositions P.1-P.7 will generally be referred to as Osmotic-controlled Release Oral Delivery System Composition.
  • the invention provides a method (Method 1) for the treatment or prophylaxis of a central nervous system disorder, comprising administering to a patient in need thereof a Compound of Formulas I et seq. or a Pharmaceutical Composition 3 or 3.1-3.4 or P.1-P.7, for example Method 1 wherein the compound or composition administered is:
  • the present disclosure provides Method 1 or any of Methods 1.1-1.5, wherein the method is further as described as follows:
  • Substance-use disorders and substance-induced disorders are the two categories of substance-related disorders defined by the Fifth Edition of the DSM (the Diagnostic and Statistical Manual of Mental Disorders.
  • a substance-use disorder is a pattern of symptoms resulting from use of a substance which the individual continues to take, despite experiencing problems as a result.
  • a substance-induced disorder is a disorder induced by use if the substance.
  • Substance-induced disorders include intoxication, withdrawal, substance induced mental disorders, including substance induced psychosis, substance induced bipolar and related disorders, substance induced depressive disorders, substance induced anxiety disorders, substance induced obsessive-compulsive and related disorders, substance induced sleep disorders, substance induced sexual dysfunctions, substance induced delirium and substance induced neurocognitive disorders.
  • the DSM-V includes criteria for classifying a substance use disorder as mild, moderate or severe.
  • the substance use disorder is selected from a mild substance use disorder, a moderate substance use disorder or a severe substance use disorder.
  • the substance use disorder is a mild substance use disorder.
  • the substance use disorder is a moderate substance use disorder.
  • the substance use disorder is a severe substance use disorder.
  • Anxiety and depression are highly prevalent co-morbid disorders in patients undergoing treatment of substance use or substance abuse.
  • a common treatment for substance abuse disorder is the combination of the partial opioid agonist buprenorphine with the opioid antagonist naloxone, but neither of these drugs has any significant effect on anxiety or depression, thus leading to the common result that a third drug, such as a benzodiazepine-class anxiolytic agent or an SSRI anti-depressant, must also be prescribed. This makes treatment regimens and patient compliance more difficult.
  • the Compounds of the present disclosure provide opiate antagonism along with serotonin antagonism and dopamine modulation. This may result in significant enhancement of treatment of patients with substance use or abuse disorder concomitant with anxiety and/or depression.
  • the compounds of the present disclosure may have anxiolytic properties ameliorating the need for treatment of a patient with an anxiolytic agent where said patients suffers from co-morbid anxiety.
  • the present disclosure provides a method according to Method 1, or any of Methods 1.1-1.23, wherein the central nervous system disorder is a substance addiction, substance use disorders and/or substance-induced disorders, or a substance abuse disorder, for example, in a patient suffering from symptoms of anxiety or who is diagnosed with anxiety as a co-morbid disorder, or as a residual disorder, wherein the method does not comprise the further administration of an anxiolytic agent, such as a benzodiazepine.
  • Benzodiazepines are GABA-modulating compounds, including those discussed with reference to Method 3.1 and 3.2 below.
  • the present disclosure provides Method 1 or any of Methods 1.1-1.7, wherein the method is further as described as follows:
  • the present disclosure provides any of the Methods 1 or 1.1-1.31 as hereinbefore described wherein the disorder is schizophrenia or sleep disorder. In some embodiments, said schizophrenia is associated with depression.
  • the present disclosure provides any of Methods 1.1-1.31, wherein the Pharmaceutical Composition 3 or 3.1-3.4, or Pharmaceutical Composition P.1-P.7, is administered for controlled- and/or sustained-release of the Compounds of the Invention over a period of from about 14 days, about 30 to about 180 days, preferably over the period of about 30, about 60 or about 90 days. Controlled- and/or sustained-release is particularly useful for circumventing premature discontinuation of therapy, particularly for antipsychotic drug therapy where non-compliance or non-adherence to medication regimes is a common occurrence.
  • the invention provides any Method 1 or 1.1-1.31 as hereinbefore described, wherein the Depot Composition of the present disclosure is administered for controlled- and/or sustained-release of the Compounds of the Invention over a period of time.
  • the invention provides a method (Method 2) for the prophylaxis or treatment of one or more sleep disorders comprising administering to a patient in need thereof a Compound of Formulas I et seq. or a Pharmaceutical Composition 3 or 3.1-3.4 or P.1-P.7, (Method 2) for example Method 2 wherein the compound or composition administered is:
  • the invention provides Method 2, or 2.1-2.5, wherein the sleep disorder includes sleep maintenance insomnia, frequent awakenings, and waking up feeling unrefreshed; for example:
  • the invention provides Method 2, or any of 2.1-2.10, wherein the sleep disorder includes sleep maintenance insomnia, frequent awakenings, and waking up feeling unrefreshed.
  • the Compounds of the present disclosure, the Pharmaceutical Compositions of the present disclosure or the Depot Compositions of the present disclosure may be used in combination with a second therapeutic agent, particularly at lower dosages than when the individual agents are used as a monotherapy so as to enhance the therapeutic activities of the combined agents without causing the undesirable side effects commonly occur in conventional monotherapy. Therefore, the Compounds of the present disclosure may be simultaneously, sequentially, or contemporaneously administered with other anti-depressant, anti-psychotic, other hypnotic agents, and/or agents use to treat Parkinson's disease or mood disorders.
  • side effects may be reduced or minimized by administering a Compound of the present disclosure in combination with one or more second therapeutic agents in free or salt form, wherein the dosages of (i) the second therapeutic agent(s) or (ii) both Compound of the present disclosure and the second therapeutic agents, are lower than if the agents/compounds are administered as a monotherapy.
  • the Compounds of the present disclosure are useful to treat dyskinesia in a patient receiving dopaminergic medications, e.g., selected from levodopa and levodopa adjuncts (carbidopa, COMT inhibitors, MAO-B inhibitors), dopamine agonists, and anticholinergics, e.g., such as are used in the treatment of Parkinson's disease.
  • dopaminergic medications e.g., selected from levodopa and levodopa adjuncts (carbidopa, COMT inhibitors, MAO-B inhibitors), dopamine agonists, and anticholinergics, e.g., such as are used in the treatment of Parkinson's disease.
  • the present disclosure provides Method I, or any of Methods 1.1-1.31, or Method 2 or any of 2.1-2.10, further comprising the administration of one or more therapeutic agents to the patient, wherein the one or more therapeutic agents is selected from compounds that modulate GABA activity (e.g., enhances the activity and facilitates GABA transmission), a GABA-B agonist, a 5-HT receptor modulator (e.g., a 5-HT 1A agonist, a 5-HT 2A antagonist, a 5-HT 2A inverse agonist, etc.), a melatonin receptor agonist, an ion channel modulator (e.g., blocker), a serotonin-2 receptor antagonist/reuptake inhibitor (a compound having both 5-HT 2 antagonism and serotonin reuptake inhibition, i.e., SARIs), an orexin receptor antagonist, an H3 agonist or antagonist, a noradrenergic agonist or antagonist, a galanin agonist, a CRH antagonist,
  • GABA activity e
  • the present disclosure provides Method I, or any of Methods 1.1-1.31, or Method 2 or any of 2.1-2.10, further comprising the administration to the patient of one or more therapeutic agents selected from the foregoing and further selected from agonists or partial agonists of the mu-opiate, kappa-opiate, delta-opiate, and/or nociceptin/orphanin receptors.
  • the present disclosure also provides Method I, or any of Methods 1.1-31, or Method 2 or any of 2.1-2.10, further comprising one or more therapeutic agents selected from a serotonin HT6 receptor antagonist, and an mGluR-2, -3 or -5 receptor agonist or antagonist (including both positive and negative modulators and partial agonists).
  • the invention provides Method 3 (i.e., Method I-A or II-A), wherein the method further comprises the administration of one or more therapeutic agents to the patient, as follows:
  • the combination of a Compound of the present disclosure and one or more second therapeutic agents as described in Methods I-A, II-A or any of Methods 3 or 3.1-3.40 may be administered to the patient as a Pharmaceutical Composition or a depot Composition as hereinbefore described.
  • the combination compositions can include mixtures of the combined drugs, as well as two or more separate compositions of the drugs, which individual compositions can be, for example, co-administered together to a patient.
  • Methods I-A, II-A, 3 or 3.1-3.40 comprises administering to a patient in need thereof, a Compound of the Invention in combination with an atypical antipsychotic agent, e.g., a compound selected from brexpiprazole, cariprazine, asenapine, lurasidone, clozapine, aripiprazole, olanzapine, quetiapine, risperidone, ziprasidone, or paliperidone, in free or pharmaceutically acceptable salt form, for example wherein the dosage of the atypical antipsychotic agent is reduced and/or side effects are reduced.
  • an atypical antipsychotic agent e.g., a compound selected from brexpiprazole, cariprazine, asenapine, lurasidone, clozapine, aripiprazole, olanzapine, quetiapine, risperidone, ziprasidone,
  • Methods I-A, II-A, 3 or 3.1-3.40 comprises administering to the patient in need thereof, a Compound of the Invention in combination with an anti-depressant, e.g., amitriptyline, amoxapine, bupropion, citalopram, clomipramine, desipramine, doxepin, duloxetine, escitalopram, fluoxetine, fluvoxamine, imipramine, isocarboxazid, maprotiline, mirtazapine, nefazodone, nortriptyline, paroxetine, phenelzine sulfate, protriptyline, sertraline, tranylcypromine, trazodone, trimipramine, or venlafaxine, in free or pharmaceutically acceptable salt form.
  • an anti-depressant e.g., amitriptyline, amoxapine, bupropion, citalopram, clomipramine,
  • Methods I-A, II-A, 3 or 3.1-3.40 comprises administering to the patient in need thereof, a Compound of the Invention in combination with a compound that modulates GABA activity, e.g., a compound selected from doxepin, alprazolam, bromazepam, clobazam, clonazepam, clorazepate, diazepam, flunitrazepam, flurazepam, lorazepam, midazolam, nitrazepam, oxazepam, temazepam, triazolam, indiplon, zopiclone, eszopiclone, zaleplon, Zolpidem, gaboxadol, vigabatrin, tiagabine, EVT 201 (Evotec Pharmaceuticals), estazolam or any combinations thereof, in free or pharmaceutically acceptable salt form.
  • the methods disclosed herein do not further comprise administration of
  • Methods I-A, II-A, 3 or 3.1-3.40 comprises administering to a patient in need thereof, a Compound of the Invention in combination with doxepin in free or pharmaceutically acceptable salt form.
  • Dosages of doxepin can vary in any range known to a person of ordinary skill in the art. In one example, a 10 mg dose of doxepin may be combined with any dosage of a compound of the Invention.
  • Methods I-A, II-A, 3 or 3.1-3.40 comprises administering to a patient in need thereof, a Compound of the Invention in combination (including as part of a daily dosage regimen) with an atypical stimulant, e.g., a modafinil, adrafinil, or armodafinil
  • an atypical stimulant e.g., a modafinil, adrafinil, or armodafinil
  • a regimen incorporating a Compound of the Invention with such drugs promotes more regular sleep, and avoids side effects such as psychosis or mania associated with higher levels of such drugs, e.g., in the treatment of bipolar depression, cognition associated with schizophrenia, and excessive sleepiness and fatigue in conditions such as Parkinson's disease and cancer.
  • each of the Compounds of Formulas I et seq.; Pharmaceutical Compositions 3 and 3.1-3.4; Compositions P.1-P.7; Methods 1 and 1.1-1.31; and Methods 2 and 2.1-2.10 and 3 and 3.1-3.40; the compound of the present disclosure is substantially free of compound of Formula A and/or Formula B.
  • the invention provides use of a compound as described in the following:
  • the invention provides a pharmaceutical composition as hereinbefore described, e.g.:
  • the current invention provides compounds which specifically limit, slow, alter and/or prevent the metabolism which has been found to occur in animals treated with the compounds such as the Compound A and Compound B:
  • deuterium ( 2 H) atoms Due to the very similar chemical and physical properties of deuterium ( 2 H) atoms compared to normal hydrogen atoms ( 1 H), e.g., atomic charge, atomic volume, polarity, valency, etc., drug compounds in which deuterium is substituted for hydrogen are believed to generally have similar biological activity to the non-deuterated analog, but potentially with improved pharmacokinetic properties. It is particularly important that while deuterium atoms have almost double the atomic mass of protium atoms, their space volume and charge distribution are similar, these latter factors being critical in binding to biological molecules.
  • deuterated pharmaceutical compounds Although many deuterated pharmaceutical compounds have been proposed and explored to date, only one deuterated pharmaceutical compound has been approved by the U.S. Food and Drug Administration, deutetrabenazine (Teva Pharmaceuticals, April 2017), a deuterated version of the Huntington's disease drug tetrabenazine, which has a therapeutically useful longer half-life than its non-deuterated counterpart.
  • the current disclosure provides compounds containing deuterium atoms at specific selected positions of the structure of compounds of Formula A and/or Formula B. These particular deuterations are expected to have in impact on metabolic degradation and clearance of said compounds because of their relationship to enzymatic pathways determined by the inventors to likely affect these compounds. These novel compounds are therefore expected to antagonize 5-HT 2A receptors, inhibit the serotonin re-uptake transporter, modulate dopaminergic protein phosphorylation, and modulate mu-opiate receptor activity, in a like manner as to their natural hydrogen analogs, yet with unexpectedly improved metabolic stability and pharmacokinetic properties.
  • Alkyl as used herein is a saturated or unsaturated hydrocarbon moiety, e.g., one to twenty-one carbon atoms in length, unless indicated otherwise; any such alkyl may be linear or branched (e.g., n-butyl or tert-butyl), preferably linear, unless otherwise specified.
  • C 1-21 alkyl denotes alkyl having 1 to 21 carbon atoms.
  • alkyl is optionally substituted with one or more hydroxy or C 1-22 alkoxy (e.g., ethoxy) groups.
  • alkyl contains 1 to 21 carbon atoms, preferably straight chain and optionally saturated or unsaturated, for example in some embodiments wherein R 1 is an alkyl chain containing 1 to 21 carbon atoms, preferably 6-15 carbon atoms, 16-21 carbon atoms, e.g., so that together with the —C(O)— to which it attaches, e.g., when cleaved from the compound of Formula I, forms the residue of a natural or unnatural, saturated or unsaturated fatty acid.
  • D refers to the 2 H-isotope of the atom hydrogen.
  • the natural abundance of the two stable isotopes of hydrogen are about 99.98% protium ( 1 H), and 0.02% deuterium ( 2 H).
  • protium 1 H
  • deuterium 2 H
  • any hydrogen atom in a molecule synthesized using common reagents will have approximately 0.02% deuterium at every hydrogen atom position.
  • a label “D” in a molecule indicates, e.g., at least 0.1% deuterium, or at least 1% deuterium, or at least 10% deuterium.
  • any compound according to the present disclosure has greater than 50% incorporation of deuterium at each specified “D” atom position of the compound's structure (i.e., greater than 50 atom % D), e.g., greater than 60%, or greater than 70%, or greater than 80%, or greater than 90% or greater than 95%, or greater than 96%, or greater than 97%, or greater than 98%, or greater than 99%.
  • pharmaceutically acceptable diluent or carrier is intended to mean diluents and carriers that are useful in pharmaceutical preparations, and that are free of substances that are allergenic, pyrogenic or pathogenic, and that are known to potentially cause or promote illness.
  • Pharmaceutically acceptable diluents or carriers thus exclude bodily fluids such as example blood, urine, spinal fluid, saliva, and the like, as well as their constituent components such as blood cells and circulating proteins.
  • Suitable pharmaceutically acceptable diluents and carriers can be found in any of several well-known treatises on pharmaceutical formulations, for example Anderson, Philip O.; Knoben, James E.; Troutman, William G, eds., Handbook of Clinical Drug Data, Tenth Edition, McGraw-Hill, 2002; Pratt and Taylor, eds., Principles of Drug Action, Third Edition, Churchill Livingston, N.Y., 1990; Katzung, ed., Basic and Clinical Pharmacology, Ninth Edition, McGraw Hill, 20037ybg; Goodman and Gilman, eds., The Pharmacological Basis of Therapeutics, Tenth Edition, McGraw Hill, 2001; Remington's Pharmaceutical Sciences, 20th Ed., Lippincott Williams & Wilkins., 2000; and Martindale, The Extra Pharmacopoeia, Thirty-Second Edition (The Pharmaceutical Press, London, 1999); all of which are incorporated by reference herein in their entirety.
  • purified refers to the physical state of said compound after being isolated from a synthetic process (e.g., from a reaction mixture), or natural source or combination thereof.
  • purified refers to the physical state of said compound after being obtained from a purification process or processes described herein or well known to the skilled artisan (e.g., chromatography, recrystallization, LC-MS and LC-MS/MS techniques and the like), in sufficient purity to be characterizable by standard analytical techniques described herein or well known to the skilled artisan.
  • an acid-addition salt of a compound of the invention which is sufficiently basic, for example, an acid-addition salt with, for example, an inorganic or organic acid, for example hydrochloric, hydrobromic, sulphuric, phosphoric, acid acetic, trifluoroacetic, citric, maleic acid, toluene sulfonic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic,
  • a salt of a compound of the invention which is sufficiently acidic is an alkali metal salt, for example a sodium or potassium salt, an alkaline earth metal salt, for example a calcium or magnesium salt, an ammonium salt or a salt with an organic base which affords a physiologically-acceptable cation, for example a salt with methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)-amine.
  • the salt of the Compounds of the Invention is a toluenesulfonic acid addition salt.
  • the Compounds of the present disclosure may comprise one or more chiral carbon atoms.
  • the compounds thus exist in individual isomeric, e.g., enantiomeric or diastereomeric form or as mixtures of individual forms, e.g., racemic/diastereomeric mixtures. Any isomer may be present in which the asymmetric center is in the (R)-, (S)-, or (R,S)-configuration.
  • the invention is to be understood as embracing both individual optically active isomers as well as mixtures (e.g., racemic/diastereomeric mixtures) thereof.
  • the Compounds of the Invention may be a racemic mixture or it may be predominantly, e.g., in pure, or substantially pure, isomeric form, e.g., greater than 70% enantiomeric/diastereomeric excess (“ee”), preferably greater than 80% ee, more preferably greater than 90% ee, most preferably greater than 95% ee.
  • ee enantiomeric/diastereomeric excess
  • the purification of said isomers and the separation of said isomeric mixtures may be accomplished by standard techniques known in the art (e.g., column chromatography, preparative TLC, preparative HPLC, simulated moving bed and the like).
  • Geometric isomers by nature of substituents about a double bond or a ring may be present in cis (Z) or trans (E) form, and both isomeric forms are encompassed within the scope of this invention.
  • the compounds of the present disclosure encompass their stable and unstable isotopes.
  • Stable isotopes are nonradioactive isotopes which contain one additional neutron compared to the abundant nuclides of the same species (i.e., element). It is expected that the activity of compounds comprising such isotopes would be retained, and such compound would also have utility for measuring pharmacokinetics of the non-isotopic analogs.
  • the hydrogen atom at a certain position on the compounds of the disclosure may be replaced with deuterium (a stable isotope which is non-radioactive). Examples of known stable isotopes include, but not limited to, deuterium, 13 C, 15 N, 18 O.
  • unstable isotopes which are radioactive isotopes which contain additional neutrons compared to the abundant nuclides of the same species (i.e., element), e.g., 123 I, 131 I, 125 I, 11 C, 18 F, may replace the corresponding abundant species of I, C and F.
  • Another example of useful isotope of the compound of the invention is the 11 C isotope.
  • the present disclosure further envisions compounds according to Formula I wherein one or more carbon atoms, nitrogen atoms or oxygen atoms are replaced by a stable or unstable isotopic variant (e.g., 11 C, 13 C 15 N, 18 O, 18 F), and further wherein one or more hydrogen atoms are replaced by tritium ( 3 H).
  • a stable or unstable isotopic variant e.g., 11 C, 13 C 15 N, 18 O, 18 F
  • one or more hydrogen atoms are replaced by tritium ( 3 H).
  • These compounds are useful, e.g., for structural determinations (e.g., by nuclear magnetic resonance or mass spectral analysis) and for the purpose of radioimaging studies to elucidate metabolic and excretory pathways and to measure clearance of potential drug candidates.
  • Compounds of the present disclosure may be included as a depot formulation, e.g., by dispersing, dissolving or encapsulating the Compounds of the Invention in a polymeric matrix as described in any of Composition 3 and 3.1-3.4, such that the Compound is continually released as the polymer degrades over time.
  • the release of the Compounds of the Invention from the polymeric matrix provides for the controlled- and/or delayed- and/or sustained-release of the Compounds, e.g., from the pharmaceutical depot composition, into a subject, for example a warm-blooded animal such as man, to which the pharmaceutical depot is administered.
  • the pharmaceutical depot delivers the Compounds of the Invention to the subject at concentrations effective for treatment of the particular disease or medical condition over a sustained period of time, e.g., 14-180 days, preferably about 30, about 60 or about 90 days.
  • Polymers useful for the polymeric matrix in the Composition of the Invention may include a polyester of a hydroxyfatty acid and derivatives thereof or other agents such as polylactic acid, polyglycolic acid, polycitric acid, polymalic acid, poly-beta.-hydroxybutyric acid, epsilon.-capro-lactone ring opening polymer, lactic acid-glycolic acid copolymer, 2-hydroxybutyric acid-glycolic acid copolymer, polylactic acid-polyethyleneglycol copolymer or polyglycolic acid-polyethyleneglycol copolymer), a polymer of an alkyl alpha-cyanoacrylate (for example poly(butyl 2-cyanoacrylate)), a polyalkylene oxalate (for example polytrimethylene oxalate or polytetramethylene oxalate), a polyortho ester, a polycarbonate (for example polyethylene carbon
  • the polymers are copolymers, they may be any of random, block and/or graft copolymers.
  • any one of D-isomers, L-isomers and/or DL-isomers may be used.
  • alpha-hydroxycarboxylic acid polymer preferably lactic acid-glycolic acid polymer
  • its ester preferably lactic acid-glycolic acid polymer
  • poly-alpha-cyanoacrylic acid esters etc.
  • lactic acid-glycolic acid copolymer also referred to as poly(lactide-alpha-glycolide) or poly(lactic-co-glycolic acid), and hereinafter referred to as PLGA
  • PLGA lactic acid-glycolic acid copolymer
  • the polymer useful for the polymeric matrix is PLGA.
  • the term PLGA includes polymers of lactic acid (also referred to as polylactide, poly(lactic acid), or PLA). Most preferably, the polymer is the biodegradable poly(d,l-lactide-co-glycolide) polymer.
  • the polymeric matrix of the invention is a biocompatible and biodegradable polymeric material.
  • biocompatible is defined as a polymeric material that is not toxic, is not carcinogenic, and does not significantly induce inflammation in body tissues.
  • the matrix material should be biodegradable wherein the polymeric material should degrade by bodily processes to products readily disposable by the body and should not accumulate in the body.
  • the products of the biodegradation should also be biocompatible with the body in that the polymeric matrix is biocompatible with the body.
  • polymeric matrix materials include poly(glycolic acid), poly-D,L-lactic acid, poly-L-lactic acid, copolymers of the foregoing, poly(aliphatic carboxylic acids), copolyoxalates, polycaprolactone, polydioxanone, poly(ortho carbonates), poly(acetals), poly(lactic acid-caprolactone), polyorthoesters, poly(glycolic acid-caprolactone), polyanhydrides, and natural polymers including albumin, casein, and waxes, such as, glycerol mono- and distearate, and the like.
  • the preferred polymer for use in the practice of this invention is dl(polylactide-co-glycolide). It is preferred that the molar ratio of lactide to glycolide in such a copolymer be in the range of from about 75:25 to 50:50.
  • Useful PLGA polymers may have a weight-average molecular weight of from about 5,000 to 500,000 Daltons, preferably about 150,000 Daltons. Dependent on the rate of degradation to be achieved, different molecular weight of polymers may be used. For a diffusional mechanism of drug release, the polymer should remain intact until all of the drug is released from the polymeric matrix and then degrade. The drug can also be released from the polymeric matrix as the polymeric excipient bioerodes.
  • the PLGA may be prepared by any conventional method, or may be commercially available.
  • PLGA can be produced by ring-opening polymerization with a suitable catalyst from cyclic lactide, glycolide, etc. (see EP-0058481B2; Effects of polymerization variables on PLGA properties: molecular weight, composition and chain structure).
  • PLGA is biodegradable by means of the degradation of the entire solid polymer composition, due to the break-down of hydrolysable and enzymatically cleavable ester linkages under biological conditions (for example in the presence of water and biological enzymes found in tissues of warm-blooded animals such as humans) to form lactic acid and glycolic acid.
  • Both lactic acid and glycolic acid are water-soluble, non-toxic products of normal metabolism, which may further biodegrade to form carbon dioxide and water.
  • PLGA is believed to degrade by means of hydrolysis of its ester groups in the presence of water, for example in the body of a warm-blooded animal such as man, to produce lactic acid and glycolic acid and create the acidic microclimate. Lactic and glycolic acid are by-products of various metabolic pathways in the body of a warm-blooded animal such as man under normal physiological conditions and therefore are well tolerated and produce minimal systemic toxicity.
  • the polymeric matrix useful for the invention may comprise a star polymer wherein the structure of the polyester is star-shaped.
  • These polyesters have a single polyol residue as a central moiety surrounded by acid residue chains.
  • the polyol moiety may be, e. g., glucose or, e. g., mannitol.
  • esters are known and described in GB 2,145,422 and in U.S. Pat. No. 5,538,739, the contents of which are incorporated by reference.
  • the star polymers may be prepared using polyhydroxy compounds, e. g., polyol, e.g., glucose or mannitol as the initiator.
  • the polyol contains at least 3 hydroxy groups and has a molecular weight of up to about 20,000 Daltons, with at least 1, preferably at least 2, e.g., as a mean 3 of the hydroxy groups of the polyol being in the form of ester groups, which contain polylactide or co-polylactide chains.
  • the branched polyesters, e.g., poly (d, 1-lactide-co-glycolide) have a central glucose moiety having rays of linear polylactide chains.
  • the depot compositions of the invention may comprise the polymer in the form of microparticles or nanoparticles, or in a liquid form, with the Compounds of the Invention dispersed or encapsulated therein.
  • “Microparticles” is meant solid particles that contain the Compounds of the Invention either in solution or in solid form wherein such compound is dispersed or dissolved within the polymer that serves as the matrix of the particle.
  • the microparticles may be prepared using any appropriate method, such as by a solvent evaporation or solvent extraction method.
  • a solvent evaporation method the Compounds of the Invention and the polymer may be dissolved in a volatile organic solvent (for example a ketone such as acetone, a halogenated hydrocarbon such as chloroform or methylene chloride, a halogenated aromatic hydrocarbon, a cyclic ether such as dioxane, an ester such as ethyl acetate, a nitrile such as acetonitrile, or an alcohol such as ethanol) and dispersed in an aqueous phase containing a suitable emulsion stabilizer (for example polyvinyl alcohol, PVA).
  • a suitable emulsion stabilizer for example polyvinyl alcohol, PVA
  • the organic solvent is then evaporated to provide microparticles with the Compounds of the Invention encapsulated therein.
  • the Compounds of the Invention and polymer may be dissolved in a polar solvent (such as acetonitrile, dichloromethane, methanol, ethyl acetate or methyl formate) and then dispersed in an aqueous phase (such as a water/PVA solution).
  • a polar solvent such as acetonitrile, dichloromethane, methanol, ethyl acetate or methyl formate
  • an aqueous phase such as a water/PVA solution
  • Spray drying is an alternative manufacturing technique for preparing the microparticles.
  • the microparticle of the present invention can be prepared by any method capable of producing microparticles in a size range acceptable for use in an injectable composition.
  • One preferred method of preparation is that described in U.S. Pat. No. 4,389,330.
  • the active agent is dissolved or dispersed in an appropriate solvent.
  • the agent-containing medium is added the polymeric matrix material in an amount relative to the active ingredient that provides a product having the desired loading of active agent.
  • all of the ingredients of the microparticle product can be blended in the solvent medium together.
  • Solvents for the Compounds of the Invention and the polymeric matrix material that can be employed in the practice of the present invention include organic solvents, such as acetone; halogenated hydrocarbons, such as chloroform, methylene chloride, and the like; aromatic hydrocarbon compounds; halogenated aromatic hydrocarbon compounds; cyclic ethers; alcohols, such as, benzyl alcohol; ethyl acetate; and the like.
  • the solvent for use in the practice of the present invention may be a mixture of benzyl alcohol and ethyl acetate.
  • the amount of the Compounds of the present disclosure incorporated in the microparticles usually ranges from about 1 wt % to about 90 wt. %, preferably 30 to 50 wt. %, more preferably 35 to 40 wt. %.
  • weight % is meant parts of the Compounds of the present disclosure per total weight of microparticle.
  • the pharmaceutical depot compositions may comprise a pharmaceutically-acceptable diluent or carrier, such as a water miscible diluent or carrier.
  • a “therapeutically effective amount” is any amount of the Compounds of the invention (for example as contained in the pharmaceutical depot) which, when administered to a subject suffering from a disease or disorder, is effective to cause a reduction, remission, or regression of the disease or disorder over the period of time as intended for the treatment.
  • an amount of the Compound of the Invention for administration refers to or is based on the amount of the Compound of the Invention in free base form (i.e., the calculation of the amount is based on the free base amount).
  • Compounds of the Invention may be administered by any satisfactory route, including orally, parenterally (intravenously, intramuscular or subcutaneous) or transdermally, but are preferably administered orally.
  • the Compounds of the Invention e.g., in depot formulation, are preferably administered parenterally, e.g., by injection.
  • Satisfactory results for Method I-A or Method II-A, or any of 3.1-3.40 are indicated to be obtained at less than 100 mg, preferably less than 50 mg, e.g., less than 40 mg, less than 30 mg, less than 20 mg, less than 10 mg, less than 5 mg, less than 2.5 mg, once daily. Satisfactory results for Method II-A or any of 3.1-3.40 are indicated to be obtained at less than 5 mg, preferably less than 2.5 mg.
  • the dosages will be higher relative to the shorter action composition, e.g., higher than 1-100 mg, e.g., 25 mg, 50 mg, 100 mg, 500 mg, 1,000 mg, or greater than 1000 mg.
  • Duration of action of the Compounds of the present disclosure may be controlled by manipulation of the polymer composition, i.e., the polymer:drug ratio and microparticle size. Wherein the composition of the invention is a depot composition, administration by injection is preferred.
  • the pharmaceutically acceptable salts of the Compounds of the present disclosure can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free base forms of these compounds with a stoichiometric amount of the appropriate acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • Further details for the preparation of these salts e.g., toluenesulfonic salt in amorphous or crystal form, may be found in PCT/US08/03340 and/or U.S. Provisional Appl. No. 61/036,069 (each equivalent to US 2011/112105).
  • compositions comprising Compounds of the present disclosure may be prepared using conventional diluents or excipients (an example include, but is not limited to sesame oil) and techniques known in the galenic art.
  • oral dosage forms may include tablets, capsules, solutions, suspensions and the like.
  • the essential core of other Compounds of the present disclosure came be made by analogous procedures disclosed in the above-reference publications and known to those skilled in the art.
  • the particular deuterated compounds of the present disclosure may generally be prepared by analogous means by substituting commercially available deuterated reagents for non-deuterated reagents, when such deuterated reagents are available.
  • Isolation or purification of the diastereomers of the Compounds of the Invention may be achieved by conventional methods known in the art, e.g., column purification, preparative thin layer chromatography, preparative HPLC, crystallization, trituration, simulated moving beds and the like.
  • Salts of the Compounds of the present disclosure may be prepared as similarly described in U.S. Pat. Nos. 6,548,493; 7,238,690; 6,552,017; 6,713,471; 7,183,282; U.S. RE39680; U.S. RE39679; and WO 2009/114181 (US 2011/112105), the contents of each of which are incorporated by reference in their entirety.
  • Diastereomers of prepared compounds can be separated by, for example, HPLC using CHIRALPAK® AY-H, 5 ⁇ , 30 ⁇ 250 mm at room temperature and eluted with 10% ethanol/90% hexane/0.1% dimethylethylamine Peaks can be detected at 230 nm to produce 98-99.9% ee of the diastereomer.
  • Step 1 To a degassed mixture of (4aS,9bR)-ethyl 6-bromo-3,4,4a,5-tetrahydro-1H-pyrido[4,3-b]indole-2(9bH)-carboxylate (1.60 g, 8.0 mmol), 2-chloro-2,2-di-deuterioacetamide (2.5 g, 26 mmol), and KI (2.68 g, 16 mmol) in dioxane (30 mL), diisopropylethylamine (3.0 mL, 16 mmol) is added at room temperature. The reaction mixture is then heated to 104° C. under vigorous stirring for 5 days.
  • Step 2 To a degassed mixture of the product from Step 1 (1.26 g, 3.3 mmol), K 2 CO 3 (1.0 g, 6.0 mmol), CuI (132 mg, 0.69 mmol) in dioxane (6 mL), and N,N,N,N′-tetramethylethylenediamine (0.3 mL, 12.0 mmol) is added at room temperature. The reaction mixture is heated to 99° C. and stirred at this temperature for 20 h. After cooling to room temperature, the mixture is directly loaded on a silica gel column.
  • the product is purified by silica gel column chromatography using 100% ethyl acetate to obtain (6bR,10aS)-ethyl 1,1-d 2 -2-oxo-2,3,6b,7,10,10 a-hexahydro-1H-pyrido [3′,4′:4,5]pyrrolo[1,2,3-de]quinoxaline-8(9H)-carboxylate as a light red solid (680 mg, yield 68%).
  • MS (ESI) m/z 318.2 [M+1]+.
  • the reaction scheme is shown below:
  • Step 3 The product from Step 2 (680 mg, 2.24 mmol) is suspended in HBr solution (33% in acetic acid, 10 ml) at room temperature. The mixture is heated to 70° C., and stirred at 70° C., for additional 2 h. LC-MS confirms the completion of reaction. The reaction mixture is cooled to room temperature and further cooled with ice. Ethyl acetate (60 mL) is added to precipitate the product salt. The solid is filtered and dried under vacuum. The HBr salt of product is suspended in methanol (20 mL) and is cooled with dry ice and 2-propanol. Ammonia (7N in methanol) is added slowly until the pH is at or above 14.
  • Step 4 A mixture of the crude product from Step 3 (227 mg, 0.98 mmol), 1-(3-chloroproxy)-4-fluorobenzene (320 ⁇ L, 2.0 mmol) and KI (330 mg, 2.0 mmol) in DMF (4 mL) is bubbled with argon for 3 minutes and DIPEA (350 ⁇ L, 2 mmol) is added. The resulting mixture is heated to 76° C. and stirred at this temperature for 2 h.
  • Step 1 To degassed CH 3 CN (5 mL), p-fluorophenol (442 mg, 4.0 mmol), 1,3-dibromopropane-d 6 (1.02 g, 4.9 mmol) and K 2 CO 3 (608 mg, 4.4 mmol) are added under stirring. The resulting mixture is heated to 80° C. and stirred at 80° C. overnight. After cooling to room temperature, the solvent is removed and the residue is suspended in dichloromethane (50 mL) and extracted with water (20 mL). The aqueous phase is separated and extracted further with dichloromethane (10 mL).
  • Step 2 A mixture of (6bR,10aS)-6b,7,8,9,10,10a-hexahydro-1H-pyrido[3′,4′:4,5]pyrrolo[1,2,3-de]quinoxalin-2(3H)-one HBr salt (900 mg, 2.9 mmol), 1-(3-bromo-1,1,2,2,3,3-d 6 -propoxy)-4-fluorobenzene (500 mg, 2.1 mmol) and potassium iodide (500 mg, 3.6 mmol) in DMF (5 mL) is bubbled with argon for 3 min, and di-isopropyl ethylamine (550 ⁇ L, 3.16 mmol) is added.
  • the resulting mixture is heated to 78° C. and stirred at this temperature for 2 h. After cooling to room temperature, the solvent is removed and the residue is suspended in dichloromethane (50 mL) and extracted with water (20 mL). The aqueous phase is separated and further extracted with dichloromethane (10 mL). The combined organic phase is dried over anhydrous Na 2 CO 3 and concentrated.
  • the final product is purified by silica gel column chromatography using a gradient of 0-80% mixed solvents [ethyl acetate/methanol/7N NH 3 (10:1:0.1 v/v)] in ethyl acetate to obtain the title product as a light brown solid (400 mg, yield 49%).
  • Step 1 To a degassed CH 3 CN (5 mL), p-fluorophenol (546 mg, 4.9 mmol), 1,3-dibromo-1,1,3,3-d 4 -propane (1.1 g, 5.4 mmol) and K 2 CO 3 (730 mg, 5.4 mmol) are added under stirring. The resulting mixture is heated to 80° C. and stirred at 80° C. for 21 h. After cooling to room temperature, the solvent is removed and the residue is suspended in dichloromethane (50 mL) and extracted with water (20 mL). The aqueous phase is separated and further extracted with dichloromethane (10 mL).
  • Step 2 A mixture of (6bR,10aS)-6b,7,8,9,10,10a-hexahydro-1H-pyrido[3′,4′:4,5]pyrrolo[1,2,3-de]quinoxalin-2(3H)-one (930 mg, 4.1 mmol), 1-(3-bromo-1,1,3,3-d 4 -propoxy)-4-fluorobenzene (500 mg, 2.1 mmol) and potassium iodide (560 mg, 3.4 mmol) in DMF (5 mL) is bubbled with argon for 3 min, and diisopropyl ethylamine (550 ⁇ L, 3.2 mmol) is added.
  • the final product is purified by silica gel column chromatography using a gradient of 0-80% mixed solvents [ethyl acetate/methanol/7N NH 3 (10:1:0.1 v/v)] in ethyl acetate and the product is further purified with basic alumina column chromatography using a gradient of 0-100% mixed solvents [ethyl acetate/methanol (20:1 v/v)] in ethyl acetate.
  • the title product is obtained as a white solid (248 mg, yield 31%).
  • the compound of Formula A and the compound of Formula B are both prepared from (6bR, 10aS)-2-oxo-2,3,6b,9,10,10a-hexahydro-1H,7H-pyrido[3′,4′:4,5]pyrrolo[1,2,3-de]quinoxaline-8-carboxylic acid ethyl ester.
  • This carbamate ester is first deprotected using HBr/acetic acid solution.
  • the resulting amine is then reacted with the appropriate alkylating agent (1-(3-chloroproxy)-4-fluorobenzene for the compound of Formula A; 4-chloro-4′-fluorobutyrophenone for the compound of Formula B) to yield the desired product.
  • Example 5 Experimental properties of 6bR,10aS)-8-(3-(4-fluorophenoxy)propyl)-6b,7,8,9,10,10a-hexahydro-1H-pyrido[3′,4′:4,5]pyrrolo[1,2,3-de]quinoxalin-2(3H)-one (the compound of Formula A) and 4-((6bR,10aS)-2-oxo-2,3,6b,9,10,10a-hexahydro-1H,7H-pyrido[3′,4′:4,5]pyrrolo[1,2,3-de]quinoxalin-8-yl)-1-(4-fluoro-phenyl)-butan-1-one (the compound of Formula B)
  • Cellular and Nuclear Receptor Functional Assays are performed on the compound of Formula B to determine the agonist and antagonist effects of the compound on the human mu-opiate receptor. It is found that the compound has an antagonist effect with an IC 50 of 1.3 ⁇ 10 ⁇ 6 M, and a KB of 1.4 ⁇ 10 ⁇ 7 M.
  • Ki (nM) or maximum inhibition Receptor Formula B Formula A 5-HT 2A 11 8.3 D2 47% inhibition at 160 240 nM D1 22 50 SERT 44% inhibition at 590 240 nM Mu opiate 22 11 receptor
  • the compounds of Formula A and B are also compared against buprenorphine (a mu-opiate partial agonist), naloxone (a mu-opiate antagonist), and DAMGO (a mu-opiate full agonist) in a both agonist and antagonist functional receptor activity assays using CHO-K1 cells expressing human mu-opiate receptor ( ⁇ 1 subtype).
  • the results are shown in Table below.
  • the results demonstrate that the compound of Formula A is a weak antagonist of the Mu receptor, showing much higher IC 50 compared to naloxone, and that it is a moderately high affinity, but partial agonist, showing only about 22% agonist activity relative to DAMGO (as compared to about 79% activity for buprenorphine relative to DAMGO).
  • the compound of Formula B is also shown to have moderately strong partial agonist activity.
  • Antagonist Agonist KB Compound IC50 (nM) EC50 (nM) (nM) Naloxone 5.80 — 0.65 DAMGO — 1.56 — Buprenorphine — 0.95 — Formula A 641 64.5 71.4 Formula B — 140 —
  • R-( ⁇ )-2,5-dimethoxy-4-iodoamphetamine is an agonist of the serotonin 5-HT 2 receptor family. When administered to mice, it produces a behavioral profile associated with frequent head twitches. The frequency of these head twitches during a predetermined period of time can be taken as an estimate of 5-HT 2 receptor agonism or antagonism in the brain.
  • DOI DOI-induced head twitch
  • the compounds of Formula A and B are also studied in a Mouse Tail Flick Assay, a measure of analgesia indicated by the pain reflex threshold of restrained mice.
  • Male CD-1 mice are positioned with their tails under a focused beam of high-intensity infrared heat source, resulting in heating of the tail.
  • the amount of time (latency) between turning on heating instrument and the flicking of the mouse's tail out of path of the heat source is recorded.
  • Administration of morphine results in analgesia, and this produces a delay in the mouse's reaction to the heat (increased latency).
  • a morphine (MOR) antagonist i.e., naloxone (NAL)
  • NAL naloxone
  • mice are a negative control, receiving vehicle both 60 minute and 30 minutes prior to the test.
  • Group 2 and 3 mice are positive controls, receiving vehicle followed by morphine or naloxone followed by morphine, respectively, prior to the test.
  • Group 4, 5 and 6 mice are the study subjects receiving the Compound of Formula A or B at one of three doses 60 minutes before the test and morphine 30 minutes before the test. The results of the study are shown below as mean tail flick latency measured in seconds:
  • mice treated with naloxone prior to treatment with the compound of Formula A at three different doses are shown in the table below as mean latency in seconds:
  • the first study demonstrates that the compounds of Formula A and B both exert a dose-dependent blockade of morphine-induced mu-opiate receptor activity.
  • the second study demonstrates that the compound of Formula A, at higher doses, exerts a dose-dependent mu-opiate agonist activity.
  • these compounds are partial agonists and partial antagonists of the mu-opiate receptor.
  • the compounds of Formula A and B are also evaluated in a mouse CNS Phosphoprotein Profile assay.
  • the extent of protein phosphorylation for selected key central nervous system proteins is measured in mice nucleus accumbens.
  • Examined proteins include ERK1, ERK2, Glul, NR2B and TH (tyrosine hydroxylase), and results are compared to the antipsychotic agents risperidone and haloperidol.
  • the results show that neither the compound of Formula A nor the compound of Formula B has a significant effect on TH phosphorylation or NR2B phosphorylation, and that they have marginal effects on GluR1 and ERK2 phosphorylation.
  • haloperidol produces a 400-500% increase in TH phosphorylation, suggesting that the compounds of Formula A and B do not disrupt dopamine metabolism.
  • the compound of Formula A is also studied in the mouse marble-burying model for OCD.
  • the marble burying test is used to measure repetitive and anxiety-related behavior in rodents. It is based on the observation that rats and mice will bury either harmful or harmless objects in their bedding, and it has been used as an animal model to measure the effect of pharmacological interventions in treatment of repetitive behavior disorders, such as OCD.
  • MPEP (2-methyl-6-(phenylethynyl)pyridine
  • mGluR5 glutamate receptor antagonist is used as a positive control.
  • Mice are administered a desired agent and placed in a cage with marbles and bedding, and after 30 minutes the number of marbles buried by the mouse is measured. The results are shown in the table below, and demonstrate that a dose-dependent reduction in OCD symptoms for the compound of Formula A.
  • the compound of Formula A is further assessed during repeated (28 day) daily subcutaneous administration to male Sprague-Dawley rats to monitor drug effects on dosing and to determine if pharmacological tolerance occurs.
  • Morphine is used as a positive control to ensure validity of the model and as a reference comparator from a similar pharmacological class.
  • the results show that repeated administration of the Compound of Example 3, at both 0.3 and 3 mg/kg four times, does not produce tolerance during subcutaneous dosing for 28 days. Furthermore, on withdrawal, a similar but decreasing profile of behavioral and physical signs is observed at the highest dose, which is not considered to be of clinical significance. Thus, overall the Compound of Example 3 was found not to produce a syndrome of physical dependence upon cessation of dosing. In contrast, repeated morphine administration is shown to produce clear signs of tolerance and dependence in this study, with changes in body weight, food and water intake, rectal temperature and clinical signs consistent with the development of tolerance and withdrawal induced dependence.
  • the Compound of Formula A is also evaluated in an oxycodone-dependent withdrawal study in mice.
  • Oxycodone is administered to mice in increasing doses over 8 days to induce physical dependent.
  • the mice are administered the compound of Formula A at one of two doses, followed either by an injection of vehicle or of naloxone.
  • the mice are then monitored for signs and symptoms of opiate withdrawal.
  • the results demonstrate that the compound of Formula A dose-dependently reduces the signs and symptoms of opiate withdrawal after the sudden cessation of opiate administration in opiate-dependent rats
  • the Compound of Formula A is also evaluated in an in the mouse formalin paw test, an inflammatory pain model.
  • Subcutaneous injection of 2.5% formalin solution into the hind paw of mice results in a biphasic response: an acute pain response and a delayed inflammatory response.
  • 30 minutes prior to formalin challenge the same paw is pre-treated with a subcutaneous injection of vehicle, morphine or the compound of Formula A in one of three doses.
  • the results demonstrate that the compound of Formula A dose-dependently attenuates both the early phase acute pain response and the late phase delayed inflammatory response to extent comparable to the morphine positive control.
  • Step 1 To a mixture of LiAlD 4 (850 mg, 20.2 mmol) in THF (25 mL) at 0° C. under vigorous stirring is added 3-(benzyloxy) propanoic acid (3.2 g, 17.7 mmol) in batches. The mixture temperature is kept under 5° C. during addition process. The mixture is then stirred at room temperature overnight and cooled to 0° C. Water (0.85 mL) and NaOH (15%, 0.85 mL) are slowly added to quench the reaction. The solvents are removed and the residue is diluted with dichloromethane (100 mL) and dried over MgSO 4 . MgSO 4 is filtered and the filtrate is evaporated to dryness. The product 3-(benzyloxy)-1,1-dideuteriopropan-1-ol is obtained as a pale solid (2.86 g, yield 96%). This product is used directly in the next step without further purification.
  • Step 2 Crude 3-(benzyloxy)-1,1-dideuteriopropan-1-ol (2.4 g, 14.3 mmol) from Step 1 is dissolved in THF (30 mL) and p-fluorophenol (1.6 g, 14.3 mmol) is added, followed by PPh 3 (3.75 g, 14.3 mmol). To this mixture under stirring, diethyl azodicarboxylate (2.3 mL, 14.6 mmol) is slowly dropped. The mixture temperature is kept within 40-50° C. during the addition process. After stirring at room temperature overnight, the reaction mixture is evaporated to dryness.
  • Step 3 Palladium on activated charcoal (10%, 0.5 g) is added to a solution of 1-(3-(benzyloxy)-1,1-dideuteriopropoxy)-4-fluorobenzene (1.89 g, 7.2 mmol) in methanol (45 mL) at room temperature. The mixture is degassed and backfilled with hydrogen three times and then stirred at room temperature for 12 h under hydrogen atmosphere. After the reaction being finished, the solid is filtered and the filtrate is evaporated to dryness. The residue is dissolved in dichloromethane (40 mL) and N, N-diisopropylethylamine (1.7 mL, 11 mmol) is added. The reaction solution is cooled to 0° C.
  • Step 4 A mixture of crude (6bR,10aS)-6b,7,8,9,10,10a-hexahydro-1H-pyrido[3′,4′:4,5]pyrrolo[1,2,3-de]quinoxalin-2(3H)-one (520 mg, 2.3 mmol) and 3,3-dideuterio-3-(4-fluorophenoxy)propyl methanesulfonate (530 mg, 2.1 mmol) in DMF (4 mL) is bubbled with argon for 3 min, and DIPEA (500 ⁇ L, 2.6 mmol) is added. The resulting mixture is heated to 78° C. and stirred at this temperature for 2 h.
  • Example 8 Synthesis of (6bR,10aS)-1,1-dideuterio-8-(1,1,2,2,3,3-hexadeuterio-3-(2,3,5,6-tetradeuterio-4-fluorophenoxy)propyl)-6b,7,8,9,10,10a-hexahydro-1H-pyrido[3′,4′:4,5]pyrrolo[1,2,3-de]quinoxalin-2(3H)-one
  • Step 1 To a degassed suspension of 4-fluorophenol (442 mg, 4.0 mmol), and K 2 CO 3 (608 mg, 4.4 mmol) in acetonitrile (5 mL) is added 1,3-dibromo-1,1,2,2,3,3-hexadeuteriopropane (1.0 g, 4.8 mmol) under stirring. The resulting mixture is heated to 80° C. and stirred at this temperature overnight. After cooling to room temperature, the reaction mixture is concentrated, and the residue is suspended in water (20 mL) and extracted with dichloromethane (2 ⁇ 30 mL). The combined dichloromethane phase is dried over Na 2 CO 3 and filtered.
  • Step 2 A mixture of (6bR,10aS)-1,1-dideuterio-6b,7,8,9,10,10a-hexahydro-1H-pyrido[3′,4′:4,5]-pyrrolo[1,2,3-de]quinoxalin-2(3H)-one HBr salt (900 mg, 2.9 mmol), 1-(3-bromo-1,1,2,2,3,3-hexadeuteriopropoxy)-4-fluorobenzene (500 mg, 2.1 mmol), and KI (500 mg, 3.6 mmol) in DMF (5 mL) is bubbled with argon for 3 min, and di-isopropyl ethylamine (500 ⁇ L, 3.16 mmol) is added.
  • the Compounds of Examples 1, 3, 6, 7, and 9, and the Compound of Formula A are tested in a radioligand binding assay using human recombinant receptors expressed in either CHO or HEK-293 cell lines.
  • the tested receptors are dopamine D 1 receptor (antagonist radioligand), dopamine D 2S receptor (agonist radioligand), mu-opioid (MOP) receptor (agonist radioligand), serotonin 5-HT 2A receptor (agonist radioligand) and serotonin transporter (SERT) (antagonist radioligand).
  • the binding assays are performed according to the procedures described in the table below:
  • the detection method is scintillation counting.
  • the results are expressed as a percentage of control specific binding ([measured specific binding]/[control specific binding]*100) and as a percent inhibition of control specific binding (100 ⁇ [[([measured specific binding]/[control specific binding]*100]).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
US17/040,892 2018-03-23 2019-03-21 Organic compounds Pending US20210009592A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/040,892 US20210009592A1 (en) 2018-03-23 2019-03-21 Organic compounds

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862647482P 2018-03-23 2018-03-23
US17/040,892 US20210009592A1 (en) 2018-03-23 2019-03-21 Organic compounds
PCT/US2019/023350 WO2019183341A1 (en) 2018-03-23 2019-03-21 Organic compounds

Publications (1)

Publication Number Publication Date
US20210009592A1 true US20210009592A1 (en) 2021-01-14

Family

ID=67987581

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/040,892 Pending US20210009592A1 (en) 2018-03-23 2019-03-21 Organic compounds
US17/040,893 Pending US20210008065A1 (en) 2018-03-23 2019-03-22 Organic compounds

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/040,893 Pending US20210008065A1 (en) 2018-03-23 2019-03-22 Organic compounds

Country Status (10)

Country Link
US (2) US20210009592A1 (es)
EP (1) EP3768271A4 (es)
JP (1) JP2021519271A (es)
CN (1) CN112088004A (es)
AU (1) AU2019236860A1 (es)
BR (1) BR112020019181A2 (es)
CA (1) CA3095101A1 (es)
IL (2) IL303857B2 (es)
MX (1) MX2020009929A (es)
WO (1) WO2019183546A1 (es)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11376249B2 (en) 2017-07-26 2022-07-05 Intra-Cellular Therapies, Inc. Organic compounds
US11753419B2 (en) 2019-12-11 2023-09-12 Intra-Cellular Therapies, Inc. 4-((6bR,10aS)-3-methyl-2,3,6b,9,10,10a-hexahydro-1H-pyrido[3′,4′:4,5]pyrrolo[1,2,3-de]quinoxalin-8(7H)-yl)-1-(4-((6bR,10aS)-3-methyl-2,3,6b,9,10,10a-hexahydro-1H-pyrido[3′4′:4,5]pyrrolo[1,2,3-de]quinoxalin-8(7H)-yl)phenyl)butan-1-one for treating conditions of the central nervous system and cardiac disorders
US12023331B2 (en) 2018-06-08 2024-07-02 Intra-Cellular Therapies, Inc. Methods

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3148303C (en) * 2013-12-03 2024-02-27 Intra-Cellular Therapies, Inc. Heterocycle fused gamma-carboline compounds for use in treatment of bipolar disorders
WO2019241278A1 (en) * 2018-06-11 2019-12-19 Intra-Cellular Therapies, Inc. Substituted heterocycle fused gamma-carbolines synthesis
WO2023225620A1 (en) 2022-05-18 2023-11-23 Intra-Cellular Therapies, Inc. Novel methods
WO2024173901A1 (en) 2023-02-17 2024-08-22 Intra-Cellular Therapies, Inc. Lumateperone and derivatives thereof for modulating the nervous system

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017117514A1 (en) * 2015-12-31 2017-07-06 Tung Roger D Deuterated iti-007
US20170319580A1 (en) * 2016-01-26 2017-11-09 Intra-Cellular Therapies, Inc. Organic compounds

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4389330A (en) 1980-10-06 1983-06-21 Stolle Research And Development Corporation Microencapsulation process
IE52535B1 (en) 1981-02-16 1987-12-09 Ici Plc Continuous release pharmaceutical compositions
US4530840A (en) 1982-07-29 1985-07-23 The Stolle Research And Development Corporation Injectable, long-acting microparticle formulation for the delivery of anti-inflammatory agents
CH656884A5 (de) 1983-08-26 1986-07-31 Sandoz Ag Polyolester, deren herstellung und verwendung.
US5538739A (en) 1989-07-07 1996-07-23 Sandoz Ltd. Sustained release formulations of water soluble peptides
WO1995013814A1 (en) 1993-11-19 1995-05-26 Janssen Pharmaceutica N.V. Microencapsulated 3-piperidinyl-substituted 1,2-benzisoxazoles and 1,2-benzisothiazoles
IL143691A0 (en) 1998-12-17 2002-04-21 Alza Corp Conversion of liquid filled gelatin capsules into controlled release systems by multiple coatings
NZ516031A (en) 1999-06-15 2003-10-31 Bristol Myers Squibb Pharma Co Substituted heterocycle fused gamma-carbolines
US6713471B1 (en) 1999-06-15 2004-03-30 Bristol-Myers Squibb Pharma Company Substituted heterocycle fused gamma-carbolines
US7071186B2 (en) 1999-06-15 2006-07-04 Bristol-Myers Squibb Pharma Co. Substituted heterocycle fused gamma-carbolines
US20050232995A1 (en) 2002-07-29 2005-10-20 Yam Nyomi V Methods and dosage forms for controlled delivery of paliperidone and risperidone
DE60316454T2 (de) 2002-07-29 2008-06-26 Alza Corp., Mountain View Verfahren und dosierformen für die kontrollierte abgabe von paliperidon
KR102372149B1 (ko) 2007-03-12 2022-03-07 인트라-셀룰라 써래피스, 인코퍼레이티드. 치환된 헤테로환 융합 감마-카르볼린 합성
AU2009223701B2 (en) 2008-03-12 2015-04-16 Intra-Cellular Therapies, Inc. Substituted heterocycle fused gamma-carbolines solid
WO2009145900A1 (en) 2008-05-27 2009-12-03 Intra-Cellular Therapies, Inc. Methods and compositions for sleep disorders and other disorders
KR101868165B1 (ko) 2010-04-22 2018-07-19 인트라-셀룰라 써래피스, 인코퍼레이티드. 유기 화합물
WO2013155506A1 (en) 2012-04-14 2013-10-17 Intra-Cellular Therapies, Inc. Novel compositions and methods
AU2013314279B2 (en) * 2012-09-14 2017-11-02 AbbVie Deutschland GmbH & Co. KG Tricyclic quinoline and quinoxaline derivatives
US9745300B2 (en) * 2014-04-04 2017-08-29 Intra-Cellular Therapies, Inc. Organic compounds
MX2021014508A (es) 2014-04-04 2023-05-18 Intra Cellular Therapies Inc Gamma-carbolinas fusionadas con heterociclos sustituidas con deuterio.
IL304192B1 (en) * 2016-03-25 2024-08-01 Intra Cellular Therapies Inc Deuterated lometaferone for use in the treatment of bipolar disorder or bipolar depression

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017117514A1 (en) * 2015-12-31 2017-07-06 Tung Roger D Deuterated iti-007
US20170319580A1 (en) * 2016-01-26 2017-11-09 Intra-Cellular Therapies, Inc. Organic compounds

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11376249B2 (en) 2017-07-26 2022-07-05 Intra-Cellular Therapies, Inc. Organic compounds
US12023331B2 (en) 2018-06-08 2024-07-02 Intra-Cellular Therapies, Inc. Methods
US11753419B2 (en) 2019-12-11 2023-09-12 Intra-Cellular Therapies, Inc. 4-((6bR,10aS)-3-methyl-2,3,6b,9,10,10a-hexahydro-1H-pyrido[3′,4′:4,5]pyrrolo[1,2,3-de]quinoxalin-8(7H)-yl)-1-(4-((6bR,10aS)-3-methyl-2,3,6b,9,10,10a-hexahydro-1H-pyrido[3′4′:4,5]pyrrolo[1,2,3-de]quinoxalin-8(7H)-yl)phenyl)butan-1-one for treating conditions of the central nervous system and cardiac disorders

Also Published As

Publication number Publication date
CN112088004A (zh) 2020-12-15
IL303857A (en) 2023-08-01
EP3768271A1 (en) 2021-01-27
IL303857B1 (en) 2024-04-01
IL277509B2 (en) 2023-11-01
JP2021519271A (ja) 2021-08-10
IL277509A (en) 2020-11-30
EP3768271A4 (en) 2021-11-17
BR112020019181A2 (pt) 2021-01-05
IL303857B2 (en) 2024-08-01
AU2019236860A1 (en) 2020-10-22
CA3095101A1 (en) 2019-09-26
WO2019183546A1 (en) 2019-09-26
US20210008065A1 (en) 2021-01-14
IL277509B1 (en) 2023-07-01
MX2020009929A (es) 2021-01-08

Similar Documents

Publication Publication Date Title
US20240165111A1 (en) Organic compounds
US11773095B2 (en) Organic compounds
US20220296591A1 (en) Organic compounds
US20210009592A1 (en) Organic compounds
US20220048910A1 (en) Organic compounds
JP2024038336A (ja) 有機化合物
US20220056031A1 (en) Organic compounds
US20220056030A1 (en) Organic compound

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: INTRA-CELLULAR THERAPIES, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LI, PENG;ZHANG, QIANG;DAVIS, ROBERT;SIGNING DATES FROM 20190328 TO 20190401;REEL/FRAME:061924/0751

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED